Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 263
Filter
1.
Microb Cell Fact ; 23(1): 259, 2024 Sep 29.
Article in English | MEDLINE | ID: mdl-39343880

ABSTRACT

BACKGROUND: Antimicrobial resistance has emerged as a major global health threat, necessitating the urgent development of new antimicrobials through innovative methods to combat the rising prevalence of resistant microbes. With this view, we developed three novel nanoconjugates using microbial natural pigment for effective application against certain pathogenic microbes. RESULTS: A natural red pigment (RP) extracted from the endophyte Monascus ruber and gamma rays were applied to synthesize RP-ZnO, RP-CuO, and RP-MgO nanoconjugates. The synthesized nanoconjugates were characterized by different techniques to study their properties. The antimicrobial potential of these nanoconjugates was evaluated. Moreover, the antibiofilm, protein leakage, growth curve, and UV light irradiation effect of the synthesized nanoconjugates were also studied. Our results confirmed the nano-size, shape, and stability of the prepared conjugates. RP-ZnO, RP-CuO, and RP-MgO nanoconjugates showed broad antimicrobial potential against the tested bacterial and fungal pathogens. Furthermore, the RP-ZnO nanoconjugate possessed the highest activity, followed by the RP-CuO against the tested microbes. The highest % inhibition of biofilm formation by the RP-ZnO nanoconjugate. Membrane leakage of E. coli and S. aureus by RP-ZnO nanoconjugate was more effective than RP-MgO and RP-CuO nanoconjugates. Finally, UV light irradiation intensified the antibiotic action of the three nanoconjugates and RP-ZnO potential was greater than that of the RP-MgO, and RP-CuO nanoconjugates. CONCLUSION: These findings pave the way for exploiting the synthesized nanoconjugates as potential materials in biomedical applications, promoting natural, green, and eco-friendly approaches.


Subject(s)
Monascus , Nanoconjugates , Monascus/metabolism , Nanoconjugates/chemistry , Biofilms/drug effects , Pigments, Biological/chemistry , Fermentation , Copper/chemistry , Copper/pharmacology , Endophytes/metabolism , Endophytes/chemistry , Zinc Oxide/chemistry , Zinc Oxide/pharmacology , Microbial Sensitivity Tests , Magnesium Oxide/chemistry , Magnesium Oxide/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Escherichia coli/drug effects
2.
Int J Nanomedicine ; 19: 9255-9271, 2024.
Article in English | MEDLINE | ID: mdl-39282577

ABSTRACT

Background: Polymeric denture materials can be susceptible to colonization by oral microorganisms. Zein-coated magnesium oxide nanoparticles (zMgO NPs) demonstrate antimicrobial activity. The aim of this study was to investigate the antimicrobial effect and adherence of different oral microorganisms on hybrid polymeric denture materials incorporated with zMgO NPs. Methods: Five types of polymeric denture materials were used. A total of 480 disc-shaped specimens were divided by material type (n=96/grp), then subdivided by zMgO NPs concentration: control with no nanoparticles and other groups with zMgO NPs concentrations of 0.3%, 0.5% and 1% by weight. Characterization of the polymeric denture materials incorporating zMgO NPs was done, and the antimicrobial activity of all groups was tested against four types of microorganisms: 1) Streptococcus mutans, 2) Staphylococcus aureus, 3) Enterococcus faecalis and 4) Candida albicans. The samples underwent an adherence test and an agar diffusion test. Experiments were done in triplicates. Results: The characterization of the hybrid samples revealed variation in the molecular composition, as well as a uniform distribution of the zMgO NPs in the polymeric denture materials. All hybrid polymeric denture materials groups induced a statistically significant antimicrobial activity, while the control groups showed the least antimicrobial activity. The agar diffusion test revealed no release of the zMgO NPs from the hybrid samples, indicating the NPs did not seep out of the matrix. Conclusion: The zMgO NPs were effective in reducing the adherence of the tested microorganisms and enhancing the antimicrobial activity of the polymeric denture materials. This antimicrobial effect with the polymeric dentures could aid in resisting microbial issues such as denture stomatitis.


Subject(s)
Anti-Infective Agents , Candida albicans , Staphylococcus aureus , Streptococcus mutans , Zein , Zein/chemistry , Zein/pharmacology , Candida albicans/drug effects , Streptococcus mutans/drug effects , Staphylococcus aureus/drug effects , Anti-Infective Agents/pharmacology , Anti-Infective Agents/chemistry , Magnesium Oxide/chemistry , Magnesium Oxide/pharmacology , Nanoparticles/chemistry , Enterococcus faecalis/drug effects , Microbial Sensitivity Tests , Humans , Dental Materials/pharmacology , Dental Materials/chemistry , Dentures/microbiology , Polymers/chemistry , Polymers/pharmacology
3.
Biomater Adv ; 164: 213977, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39094444

ABSTRACT

Biodegradable polymer microspheres in bone tissue engineering have become appealing as their non-invasive advantages in irregular damage bone repair. However, current microspheres used in BTE still lack sufficient osteogenic capacity to induce effective bone regeneration. In this study, we developed osteogenic composite microspheres concurrently loaded with magnesium oxide (MgO) and zinc oxide (ZnO), both of which are osteogenic active substances, using a facile and scalable emulsification method. The osteogenic composite microspheres exhibited a sequential yet complementary release profile characterized by a rapid release of Mg2+ and a gradual release of Zn2+ in a physiological environment, thereby maintaining the concentration of bioactive ions at a sustained high level. As a result, the combination of Mg2+ and Zn2+ in the composite microspheres led to a synergistic enhancement in biomimetic mineralization and the upregulation in the expression of osteogenic-related genes and proteins at the cellular level. Through a critical-sized calvarial rate defect model, the osteogenic composite microspheres were demonstrated to have strong osteogenic ability to promote new bone formation via ultrasonic imaging, histological and immunohistochemical evaluations. In sum, these osteogenic composite microspheres as microcarriers of Mg2+ and Zn2+ have great potential in the delivery of therapeutic ions for treating bone defects.


Subject(s)
Bone Regeneration , Magnesium , Microspheres , Osteogenesis , Bone Regeneration/drug effects , Osteogenesis/drug effects , Animals , Magnesium/pharmacology , Zinc/pharmacology , Zinc/administration & dosage , Zinc/chemistry , Zinc Oxide/pharmacology , Zinc Oxide/chemistry , Zinc Oxide/administration & dosage , Magnesium Oxide/pharmacology , Magnesium Oxide/chemistry , Magnesium Oxide/administration & dosage , Tissue Engineering/methods , Biocompatible Materials/pharmacology , Mice
4.
Sci Rep ; 14(1): 19725, 2024 08 25.
Article in English | MEDLINE | ID: mdl-39183238

ABSTRACT

Allogeneic bone grafts are used to treat bone defects in orthopedic surgery, but the osteogenic potential of artificial bones remains a challenge. In this study, we developed a ß-tricalcium phosphate (ß-TCP) formulation containing MgO, ZnO, SrO, and SiO2 and compared its bone-forming ability with that of ß-TCP without biological elements. We prepared ß-TCP discs with 60% porosity containing 1.0 wt% of these biological elements. ß-TCP scaffolds were loaded with bone marrow-derived mesenchymal stem cells (BMSC) from 7-week-old male rats and cultured for 2 weeks. ALP activity and mRNA expression of osteogenic markers were evaluated. In addition, scaffolds were implanted subcutaneously in rats and analyzed after 7 weeks. In vitro, the MgO group showed lower Ca concentrations and higher osteogenic marker expression compared to controls. In vivo, the MgO group showed higher ALP activity compared to controls, and RT-qPCR analysis showed significant expression of BMP2 and VEGF. Histopathology, fluorescent immunostaining, and micro-CT also showed relatively better bone formation in the MgO group. ß-TCP with MgO may enhance bone morphology in vitro and in vivo and improve the prognosis of patients with substantial and refractory bone defects.


Subject(s)
Calcium Phosphates , Magnesium Oxide , Mesenchymal Stem Cells , Osteogenesis , Tissue Scaffolds , Animals , Osteogenesis/drug effects , Calcium Phosphates/pharmacology , Calcium Phosphates/chemistry , Rats , Male , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Magnesium Oxide/pharmacology , Magnesium Oxide/chemistry , Tissue Scaffolds/chemistry , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 2/genetics , X-Ray Microtomography , Rats, Sprague-Dawley , Alkaline Phosphatase/metabolism , Cells, Cultured , Tissue Engineering/methods , Cell Differentiation/drug effects , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics
5.
ACS Appl Mater Interfaces ; 16(30): 39035-39050, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39026394

ABSTRACT

Given the widespread clinical demand, addressing irregular cranial bone defects poses a significant challenge following surgical procedures and traumatic events. In situ-formed injectable hydrogels are attractive for irregular bone defects due to their ease of administration and the ability to incorporate ceramics, ions, and proteins into the hydrogel. In this study, a multifunctional hydrogel composed of oxidized sodium alginate (OSA)-grafted dopamine (DO), carboxymethyl chitosan (CMCS), calcium ions (Ca2+), nanohydroxyapatite (nHA), and magnesium oxide (MgO) (DOCMCHM) was prepared to address irregular cranial bone defects via dynamic Schiff base and chelation reactions. DOCMCHM hydrogel exhibits strong adhesion to wet tissues, self-healing properties, and antibacterial characteristics. Biological evaluations indicate that DOCMCHM hydrogel has good biocompatibility, in vivo degradability, and the ability to promote cell proliferation. Importantly, DOCMCHM hydrogel, containing MgO, promotes the expression of osteogenic protein markers COL-1, OCN, and RUNX2, and stimulates the formation of new blood vessels by upregulating CD31. This study could provide meaningful insights into ion therapy for the repair of cranial bone defects.


Subject(s)
Alginates , Anti-Bacterial Agents , Chitosan , Hydrogels , Skull , Hydrogels/chemistry , Hydrogels/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Chitosan/chemistry , Chitosan/analogs & derivatives , Chitosan/pharmacology , Animals , Alginates/chemistry , Skull/drug effects , Skull/pathology , Skull/diagnostic imaging , Skull/injuries , Magnesium Oxide/chemistry , Magnesium Oxide/pharmacology , Bone Regeneration/drug effects , Dopamine/chemistry , Dopamine/pharmacology , Durapatite/chemistry , Durapatite/pharmacology , Mice , Cell Proliferation/drug effects , Calcium/metabolism , Calcium/chemistry , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Osteogenesis/drug effects , Staphylococcus aureus/drug effects
6.
Int J Biol Macromol ; 276(Pt 1): 133693, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38971277

ABSTRACT

The development of new polymer nanocomposites or antibacterial coatings is crucial in combating drug-resistant infections, particularly bacterial infections. In this study, a new chitosan polymer based nanocomposite reinforced with magnesium oxide nanopowders and carbon quantum dots was fabricated by sol-gel technique and coated on 316 L stainless steel. In order to gaining the optimal amount of components to achieve the maximum antibacterial properties, the effect of concentration of nanocomposite components on its antibacterial properties was investigated. Crystal structure, microstructure, elemental dispersion, size distribution, chemical composition and morphology of nanocomposite and coating were characterized with various analyses. The obtained results exhibited that the carbon quantum dot and magnesium oxide nanopowders were distributed uniformly and without agglomeration in the chitosan matrix and created a uniform coating. The antibacterial properties of the synthesized samples against Staphylococcus aureus bacteria (gram positive) were evaluated using disk diffusion and minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) antibacterial tests. The inhibition growth zone formed around the antibiotic and nanocomposite 25 mg/ml under dark and light was about 32 and 14, 11 mm, respectively. Also, MIC and MBC values for final nanocomposite were 62.5 and 125 µg/ml, respectively.


Subject(s)
Anti-Bacterial Agents , Chitosan , Magnesium Oxide , Microbial Sensitivity Tests , Nanocomposites , Staphylococcus aureus , Chitosan/chemistry , Chitosan/pharmacology , Nanocomposites/chemistry , Magnesium Oxide/chemistry , Magnesium Oxide/pharmacology , Staphylococcus aureus/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Quantum Dots/chemistry , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology
7.
J Biomater Sci Polym Ed ; 35(13): 1963-1977, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38949409

ABSTRACT

The interest in wound dressings increased ten years ago. Wound care practitioners can now use interactive/bioactive dressings and tissue-engineered skin substitutes. Several bandages can heal burns, but none can treat all chronic wounds. This study formulates a composite material from 70% polyvinyl alcohol (PVA) and 30% polyethylene glycol (PEG) with 0.2, 0.4, and 0.6 wt% magnesium oxide nanoparticles. This study aims to create a biodegradable wound dressing. A Fourier Transform Infrared (FTIR) study shows that PVA, PEG, and MgO create hydrogen bonding interactions. Hydrophilic characteristics are shown by the polymeric blend's 56.289° contact angle. MgO also lowers the contact angle, making the film more hydrophilic. Hydrophilicity improves film biocompatibility, live cell adhesion, wound healing, and wound dressing degradability. Differential Scanning Calorimeter (DSC) findings suggest the PVA/PEG combination melted at 53.16 °C. However, adding different weight fractions of MgO nanoparticles increased the nanocomposite's melting temperature (Tm). These nanoparticles improve the film's thermal stability, increasing Tm. In addition, MgO nanoparticles in the polymer blend increased tensile strength and elastic modulus. This is due to the blend's strong adherence to the reinforcing phase and MgO nanoparticles' ceramic material which has a great mechanical strength. The combination of 70% PVA + 30% PEG exhibited good antibacterial spatially at 0.2% MgO, according to antibacterial test results.


Subject(s)
Magnesium Oxide , Nanoparticles , Polyethylene Glycols , Polyvinyl Alcohol , Wound Healing , Polyvinyl Alcohol/chemistry , Magnesium Oxide/chemistry , Magnesium Oxide/pharmacology , Polyethylene Glycols/chemistry , Wound Healing/drug effects , Nanoparticles/chemistry , Membranes, Artificial , Tensile Strength , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Humans , Hydrophobic and Hydrophilic Interactions , Animals , Bandages , Cell Adhesion/drug effects , Materials Testing , Mice , Nanocomposites/chemistry
8.
ACS Biomater Sci Eng ; 10(8): 5226-5236, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-38943566

ABSTRACT

Metal peroxide nanomaterials as efficient hydrogen peroxide (H2O2) self-supplying agents have attracted the attention of researchers for antitumor treatment. However, relying solely on metal peroxides to provide H2O2 is undoubtedly insufficient to achieve optimal antitumor effects. Herein, we construct novel hyaluronic acid (HA)-modified nanocomposites (MgO2/Pd@HA NCs) formed by decorating palladium nanoparticles (Pd NPs) onto the surfaces of a magnesium peroxide (MgO2) nanoflower as a highly effective nanoplatform for the tumor microenvironment (TME)-responsive induction of ferroptosis in tumor cells and tumor photothermal therapy (PTT). MgO2/Pd@HA NC could be well endocytosed into tumor cells with CD44 expression depending on the specific recognition of HA with CD44, and then, the nanocomposites can be rapidly decomposed in mild acid and hyaluronidase overexpressed TME, and plenty of H2O2 was released. Simultaneously, Pd NPs catalyze self-supplied H2O2 to generate abundant hydroxyl radicals (•OH) and catalyze glutathione (GSH) into glutathione disulfide owing to its peroxidase and glutathione oxidase mimic enzyme activities, while the abundant •OH could also consume GSH in tumor cells and disturb the defense pathways of ferroptosis leading to the accumulation of lipid peroxidation and resulting in the occurrence of ferroptosis. Additionally, the superior photothermal conversion performance of Pd NPs in near-infrared II could also be used for PTT, synergistically cooperating with nanocomposite-induced ferroptosis for tumor inhibition. Consequently, the successfully prepared TME-responsive MgO2/Pd@HA NCs exhibited marked antitumor effect without obvious biotoxicity, contributing to thoroughly explore the nanocomposites as a novel and promising treatment for tumor therapy.


Subject(s)
Ferroptosis , Hyaluronic Acid , Magnesium Oxide , Nanocomposites , Palladium , Photothermal Therapy , Tumor Microenvironment , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Ferroptosis/drug effects , Tumor Microenvironment/drug effects , Magnesium Oxide/chemistry , Magnesium Oxide/pharmacology , Nanocomposites/chemistry , Nanocomposites/therapeutic use , Photothermal Therapy/methods , Animals , Humans , Palladium/chemistry , Palladium/pharmacology , Palladium/therapeutic use , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Hydrogen Peroxide
9.
Ecotoxicol Environ Saf ; 280: 116522, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38843743

ABSTRACT

This study aimed to evaluate the effect of adding liquid extract of algae (Hypnea musciformis, Grateloupia acuminata, and Sargassum muticum) (HGS) and Magnesium oxide nanoparticles (MgO NPs) using this extract to rear water of Oreochromis niloticus, on improving culture water indices, growth performance, digestive enzyme, hemato-biochemical characters, immune, antioxidative responses, and resistance after challenged by Aeromonas hydrophila with specific refer to the potential role of the mixture in vitro as resistance against three strains bacteria (Aeromonas sobria, Pseudomonas fluorescens, P. aeruginosa) and one parasite (Cichlidogyrus tilapia). The first group represented control, HGS0, whereas the other group, HGS5, HGS10, and HGS15 mL-1 of liquid extract, as well as all groups with 7.5 µg mL-1 MgO-NPs added to culture water of O. niloticus, for 60 days. Data showed that increasing levels at HGS 10 and HGS15 mL-1 in to-culture water significantly enhanced growth-stimulating digestive enzyme activity and a significantly improved survival rate of O. niloticus after being challenged with A. hydrophila than in the control group. The total viability, coliform, fecal coliform count, and heavy metal in muscle partially decreased at HGS 10 and HGS15 mL-1 than in the control group. Correspondingly, the highest positive effect on hemato-biochemical indices was noticed at levels HGS 10 and HGS15 mL-1. Fish noticed an improvement in immune and antioxidant indices compared to control groups partially at HGS 10 and HGS15 mL-1. Interestingly, fish cultured in rearing water with the mixture provided downregulated the related inflammatory genes (HSP70, TNF, IL-1ß, and IL-8) partially at HGS15 mL-1. In vitro, the mixture showed positive efficiency as an antibacterial and partially antiparasitic at HGS 10 and HGS15 mL-1. This study proposes utilizing a mixture of (HGS) and (MgO-NPs) with optimum levels of 10-15 mL-1 in cultured water to improve water indices, growth, health status, and increased resistance of O. niloticus against bacterial and parasitic infection.


Subject(s)
Cichlids , Disease Resistance , Magnesium Oxide , Water Quality , Animals , Magnesium Oxide/pharmacology , Cichlids/immunology , Disease Resistance/drug effects , Seaweed , Fish Diseases/microbiology , Fish Diseases/drug therapy , Plant Extracts/pharmacology , Plant Extracts/chemistry , Nanoparticles , Green Chemistry Technology , Metal Nanoparticles/toxicity , Metal Nanoparticles/chemistry , Aeromonas hydrophila/drug effects , Sargassum
10.
Int J Biol Macromol ; 266(Pt 1): 130995, 2024 May.
Article in English | MEDLINE | ID: mdl-38521323

ABSTRACT

Critical-size bone defects are one of the main challenges in bone tissue regeneration that determines the need to use angiogenic and osteogenic agents. Rosuvastatin (RSV) is a class of cholesterol-lowering drugs with osteogenic potential. Magnesium oxide (MgO) is an angiogenesis component affecting apatite formation. This study aims to evaluate 3D-printed Polycaprolactone/ß-tricalcium phosphate/nano-hydroxyapatite/ MgO (PCL/ß-TCP/nHA/MgO) scaffolds as a carrier for MgO and RSV in bone regeneration. For this purpose, PCL/ß-TCP/nHA/MgO scaffolds were fabricated with a 3D-printing method and coated with gelatin and RSV. The biocompatibility and osteogenicity of scaffolds were examined with MTT, ALP, and Alizarin red staining. Finally, the scaffolds were implanted in a bone defect of rat's calvaria, and tissue regeneration was investigated after 3 months. Our results showed that the simultaneous presence of RSV and MgO improved biocompatibility, wettability, degradation rate, and ALP activity but decreased mechanical strength. PCL/ß-TCP/nHA/MgO/gelatin-RSV scaffolds produced sustained release of MgO and RSV within 30 days. CT images showed that PCL/ß-TCP/nHA/MgO/gelatin-RSV scaffolds filled approximately 86.83 + 4.9 % of the defects within 3 months and improved angiogenesis, woven bone, and osteogenic genes expression. These results indicate the potential of PCL/ß-TCP/nHA/MgO/gelatin-RSV scaffolds as a promising tool for bone regeneration and clinical trials.


Subject(s)
Bone Regeneration , Gelatin , Magnesium Oxide , Osteogenesis , Printing, Three-Dimensional , Rosuvastatin Calcium , Tissue Scaffolds , Bone Regeneration/drug effects , Rosuvastatin Calcium/pharmacology , Rosuvastatin Calcium/chemistry , Tissue Scaffolds/chemistry , Gelatin/chemistry , Animals , Rats , Osteogenesis/drug effects , Magnesium Oxide/chemistry , Magnesium Oxide/pharmacology , Polyesters/chemistry , Drug Liberation , Durapatite/chemistry , Durapatite/pharmacology , Delayed-Action Preparations/pharmacology , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacology , Skull/drug effects , Tissue Engineering/methods
11.
Sci Adv ; 10(10): eadk6084, 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38457498

ABSTRACT

The emerging therapeutic strategies for osteoarthritis (OA) are shifting toward comprehensive approaches that target periarticular tissues, involving both cartilage and subchondral bone. This shift drives the development of single-component therapeutics capable of acting on multiple tissues and cells. Magnesium, an element essential for maintaining skeletal health, shows promise in treating OA. However, the precise effects of magnesium on cartilage and subchondral bone are not yet clear. Here, we investigated the therapeutic effect of Mg2+ on OA, unveiling its protective effects on both cartilage and bone at the cellular and animal levels. The beneficial effect on the cartilage-bone interaction is primarily mediated by the PI3K/AKT pathway. In addition, we developed poly(lactic-co-glycolic acid) (PLGA) microspheres loaded with nano-magnesium oxide modified with stearic acid (SA), MgO&SA@PLGA, for intra-articular injection. These microspheres demonstrated remarkable efficacy in alleviating OA in rat models, highlighting their translational potential in clinical applications.


Subject(s)
Cartilage, Articular , Nanoparticles , Osteoarthritis , Rats , Animals , Magnesium Oxide/pharmacology , Magnesium/pharmacology , Phosphatidylinositol 3-Kinases , Osteoarthritis/drug therapy
12.
Biotechnol Lett ; 46(2): 263-278, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38326543

ABSTRACT

PURPOSE: Currently, regenerative endodontic treatments are gaining more and more attention, and stem cells play a significant role in these treatments. In order to enhance stem cell proliferation and differentiation, a variety of methods and materials have been used. The purpose of this study was to determine the effects of magnesium oxide nanoparticles and LED irradiation on the survival and differentiation of human stem cells from apical papilla. METHODS: The MTT test was used to measure the cell survival of SCAPs that had been exposed to different concentrations of magnesium oxide nanoparticles after 24 and 48 h, and the concentration with the highest cell survival rate was picked for further studies. The cells were classified into four distinct groups based on their treatment: (1) control, which received no exposure, (2) exposure to magnesium oxide nanoparticles, (3) exposure to light emitting diode (LED) irradiation (635 nm, 200 mW/cm2) for 30 s, (4) exposure simultaneously with magnesium oxide nanoparticles and LED irradiation. A green approach was employed to synthesize magnesium oxide nanoparticles. Quantitative real time PCR was used to measure the gene expression of osteo/odontogenic markers such as BSP, DSPP, ALP and DMP1 in all four groups after treatment, and Alizarin red S staining (ARS) was used to determine the osteogenic differentiation of SCAPs by demonstrating the Matrix mineralization. RESULTS: The highest viability of SCAPs was observed after 24 h in concentration 1 and 10 µg/mL and after 48 h in concentration 1 µg/mL, which were not significantly different from the control group. In both times, the survival of SCAPs decreased with increasing concentration of magnesium oxide nanoparticles (MgONPs). According to the results of Real-time PCR, after 24 and 48 h, the highest differentiation of BSP, DMP1, ALP and DSPP genes was observed in the LED + MgONPs group, followed by MgONPs and then LED, and in all 3 experimental groups, it was significantly higher than control group (P < 0.05). Also, after 24 and 48 h, the density of ARS increased in all groups compared to the control group, and the highest density was observed in the MgONPs + LED and MgONPs groups. CONCLUSION: This research concluded that exposure to SCAPs, MgONPs, and LED irradiation has a significant effect on enhancing gene expression of odontogenic/osteogenic markers and increasing matrix mineralization.


Subject(s)
Magnesium Oxide , Osteogenesis , Humans , Magnesium Oxide/pharmacology , Magnesium Oxide/metabolism , Cell Differentiation , Stem Cells/metabolism , Cells, Cultured , Cell Proliferation
13.
ACS Biomater Sci Eng ; 10(3): 1676-1685, 2024 03 11.
Article in English | MEDLINE | ID: mdl-38386843

ABSTRACT

Regenerating bone tissue in critical-sized craniofacial bone defects remains challenging and requires the implementation of innovative bone implants with early stage osteogenesis and blood vessel formation. Vitamin D3 is incorporated into MgO-doped 3D-printed scaffolds for defect-specific and patient-specific implants in low load-bearing areas. This novel bone implant also promotes early stage osteogenesis and blood vessel development. Our results show that vitamin D3-loaded MgO-doped 3D-printed scaffolds enhance osteoblast cell proliferation 1.3-fold after being cultured for 7 days. Coculture studies on osteoblasts derived from human mesenchymal stem cells (hMSCs) and osteoclasts derived from monocytes show the upregulation of genes related to osteoblastogenesis and the downregulation of RANK-L, which is essential for osteoclastogenesis. Release of vitamin D3 also inhibits osteoclast differentiation by 1.9-fold after a 21-day culture. After 6 weeks, vitamin D3 release from MgO-doped 3D-printed scaffolds enhances the new bone formation, mineralization, and angiogenic potential. The multifunctional 3D-printed scaffolds can improve early stage osteogenesis and blood vessel formation in craniofacial bone defects.


Subject(s)
Magnesium Oxide , Tissue Scaffolds , Humans , Magnesium Oxide/pharmacology , Cholecalciferol/pharmacology , Printing, Three-Dimensional , Bone Regeneration
14.
Mol Pain ; 20: 17448069241233744, 2024.
Article in English | MEDLINE | ID: mdl-38323375

ABSTRACT

Methylglyoxal (MGO), a highly reactive dicarbonyl metabolite of glucose primarily formed during the glycolytic pathway, is a precursor of advanced glycation end-products (AGEs). Recently, numerous studies have shown that MGO accumulation can cause pain and hyperalgesia. However, the mechanism through which MGO induces pain in the spinal dorsal horn remains unclear. The present study investigated the effect of MGO on spontaneous excitatory postsynaptic currents (sEPSC) in rat spinal dorsal horn neurons using blind whole-cell patch-clamp recording. Perfusion of MGO increased the frequency and amplitude of sEPSC in spinal horn neurons in a concentration-dependent manner. Additionally, MGO administration increased the number of miniature EPSC (mEPSC) in the presence of tetrodotoxin, a sodium channel blocker. However, 6-cyano-7-nitroqiunocaline-2,3-dione (CNQX), an AMPA/kainate receptor antagonist, blocked the enhancement of sEPSC by MGO. HC-030031, a TRP ankyrin-1 (TRPA1) antagonist, and capsazepine, a TRP vanilloid-1 (TRPV1) antagonist, inhibited the action of MGO. Notably, the effects of MGO were completely inhibited by HC-030031 and capsazepine. MGO generates reactive oxygen species (ROS) via AGEs. ROS also potentially induce pain via TRPA1 and TRPV1 in the spinal dorsal horn. Furthermore, we examined the effect of MGO in the presence of N-tert-butyl-α-phenylnitrone (PBN), a non-selective ROS scavenger, and found that the effect of MGO was completely inhibited. These results suggest that MGO increases spontaneous glutamate release from the presynaptic terminal to spinal dorsal horn neurons through TRPA1, TRPV1, and ROS and could enhance excitatory synaptic transmission.


Subject(s)
Acetanilides , Capsaicin/analogs & derivatives , Magnesium Oxide , Purines , Pyruvaldehyde , Rats , Animals , Reactive Oxygen Species/metabolism , Pyruvaldehyde/pharmacology , Pyruvaldehyde/metabolism , Rats, Sprague-Dawley , Magnesium Oxide/metabolism , Magnesium Oxide/pharmacology , Spinal Cord Dorsal Horn/metabolism , Posterior Horn Cells/metabolism , Pain/metabolism , Synaptic Transmission/physiology
15.
Environ Sci Pollut Res Int ; 31(8): 12446-12466, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38231326

ABSTRACT

Magnesium oxide nanoparticles (MgO NPs) have great potential to enhance the crop productivity and sustainability of agriculture. Still, a thorough understanding is lacking about its essentiality or toxicity and precise dose for the safe cultivation of oilseed crops. Thus, we assessed the dual effects of MgO NPs (control, 5, 10, 20, 40, 80, and 200 mg/L) on the seed germination, growth performance, photosynthesis, total soluble protein, total carbohydrates, oxidative stress markers (hydrogen peroxide as H2O2 and superoxide anion as O2•‒), lipid peroxidation as MDA, and antioxidant defence machinery (SOD, CAT, APX, and GR activities, and GSH levels) of seven different oilseeds (Brassica napus L.) cultivars (ZY 758, ZD 649, ZD 635, ZD 619, GY 605, ZD 622, and ZD 630). Our findings revealed that low doses of MgO NPs (mainly at 10 mg/L) markedly boosted the seed germination, plant growth (shoot and root lengths) (15‒22%), and biomass (fresh and dry) (11‒19%) by improving the levels of photosynthetic pigments (14‒27%), net photosynthetic rate, stomatal conductance, photosynthetic efficiency (Fv/Fm), total soluble protein and total carbohydrates (16‒36%), antioxidant defence, and reducing the oxidative stress in B. napus tissues. Among all B. napus cultivars, these beneficial effects of MgO NPs were pronounced in ZD 635. ile, elevated levels of MgO NPs (particularly at 200 mg/L) induced oxidative stress, impaired antioxidant scavenging potential, photosynthetic inhibition, protein oxidation, and carbohydrate degradation and lead to inhibit the plant growth attributes. These inhibitory effects were more pronounced in ZD 622. Collectively, low-dose MgO NPs reinforced the Mg contents, protected the plant growth, photosynthesis, total soluble carbohydrates, enzyme activities, and minimized the oxidative stress. While, the excessive MgO NP levels impaired the above-reported traits. Overall, ZD 622 was highly susceptible to MgO NP toxicity and ZD 635 was found most tolerant to MgO NP toxicity.


Subject(s)
Brassica napus , Nanoparticles , Antioxidants/metabolism , Magnesium Oxide/pharmacology , Hydrogen Peroxide/metabolism , Oxidative Stress , Carbohydrates
16.
Plant Physiol Biochem ; 207: 108383, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38286092

ABSTRACT

Underground vegetables are sensitive and vulnerable to salt stress. The vegetables are the main source of vitamins, nutrients and minerals in human diet. Also contain healthy carbohydrates, antioxidant and resistant starch which are beneficial for human health. Salinity influences water balance, morphological appearance and cellular interference of crop plants. It also caused disproportion of nutrients which usually affects the physiochemical processes in plant. Salt stress also affect biochemical attributes and hampers the growth of underground organs, due to which yield of crop decreased. The nanoparticles had been potentially used for better crop yield, in the recent. In our research study, we elaborate the positive response of magnesium oxide nanoparticles (MgO-NPs) on the morphological and biochemical parameters as well as anti-oxidant enzymes action on two accessions of carrot (Daucus carota L.) under salt stress of 40 mM and 80 mM. In a pilot experiment, various levels (0, 50, 100, 150, 200 and 250 mg/L) of MgO-NPs were tested through foliar application on carrot plants. Foliar application of MgO-NPs at concentration of 150 mg/L was most effective treatment and ameliorate the salt stress in both carrot accessions (DC-03 and DC-90). The MgO-NPs significantly enhanced the morphological and biochemical parameters. The yield was significantly increased with the exposure of MgO-NPs. Our results thus confirmed the potential of MgO-NPs to endorse the plant development and growth under salinity. However, further research study is needed to explore effectiveness of MgO-NPs in various other plants for the ameliorant of salinity.


Subject(s)
Daucus carota , Nanoparticles , Humans , Magnesium/pharmacology , Magnesium Oxide/pharmacology , Antioxidants/pharmacology , Salt Stress
17.
Dent Mater J ; 43(1): 11-19, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38072414

ABSTRACT

Functional nano-fillers are commonly used to reduce bacterial colonization in dentistry. This study aimed to synthesize, characterize, and evaluate the biological effects of magnesium oxide (MgO) nanoparticles (NP) obtained by mechanosynthesis. XRD, TEM, FT-IR, and UV-Vis were used to characterize MgO-NP which were subsequently tested for their activity against Staphylococcus aureus, Enterococcus faecalis and Escherichia coli (E. coli). The effects of MgO-NP on osteoblast cells were also analyzed. Three variables were studied: microbial inhibition by optical density (OD; 570-nm), viability estimated by colony-forming-units, and cell proliferation. The characterization of NP is consistent with nanostructures, minimum inhibitory concentration between 1.5-5 mg/mL, and microbial inhibition at 9.75 ug/mL concentration for E. coli were determined. There were different concentration-dependent effects on cell proliferation. Results were observed with 0.156 mg/mL MgO-NP, which increased cell proliferation at 24 and 48 h. The results suggest the antibacterial suitability of MgO-NP, with tolerable viability of mammalian cells for dental applications.


Subject(s)
Magnesium Oxide , Nanoparticles , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Escherichia coli , Magnesium Oxide/pharmacology , Mammals , Microbial Sensitivity Tests , Nanoparticles/chemistry , Oxides , Spectroscopy, Fourier Transform Infrared
18.
ACS Biomater Sci Eng ; 10(1): 468-481, 2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38078836

ABSTRACT

Developing biomaterial scaffolds using tissue engineering with physical and chemical surface modification processes can improve the bioactivity and biocompatibility of the materials. The appropriate substrate and site for cell attachment are crucial in cell behavior and biological activities. Therefore, the study aims to develop a conventional electrospun nanofibrous biomaterial using reproducible surface topography, which offers beneficial effects on the cell activities of bone cells. The bioactive MgO/gC3N4 was incorporated on PAN/PEG and fabricated into a nanofibrous membrane using electrospinning. The nanocomposite uniformly distributed on the PAN/PEG nanofiber helps to increase the number of induced pores and reduce the hydrophobicity of PAN. The physiochemical characterization of prepared nanoparticles and nanofibers was carried out using FTIR, X-ray diffraction (XRD), thermogravimetry analysis (TGA), X-ray photoelectron spectroscopy (XPS), and water contact angle measurements. SEM and TEM analyses examined the nanofibrous morphology and the structure of MgO/gC3N4. In vitro studies such as on ALP activity demonstrated the membrane's ability to regenerate new bone and healing capacity. Furthermore, alizarin red staining showed the increasing ability of the cell-cell interaction and calcium content for tissue regeneration. The cytotoxicity of the prepared membrane was about 97.09% of live THP-1 cells on the surface of the MgO/gC3N4@PAN/PEG membrane evaluated using MTT dye staining. The soil burial degradation analysis exhibited that the maximum degradation occurs on the 45th day because of microbial activity. In vitro PBS degradation was observed on the 15th day after the bulk hydrolysis mechanism. Hence, on the basis of the study outcomes, we affirm that the MgO/gC3N4@PAN/PEG nanofibrous membrane can act as a potential bone regenerative substrate.


Subject(s)
Nanocomposites , Nanofibers , Tissue Scaffolds/chemistry , Nanofibers/chemistry , Magnesium Oxide/pharmacology , Biocompatible Materials , Bone Regeneration
19.
Biochem Pharmacol ; 219: 115976, 2024 01.
Article in English | MEDLINE | ID: mdl-38081372

ABSTRACT

Diabetic patients develop coronary microvascular dysfunction (CMD) and exhibit high mortality of coronary artery disease. Methylglyoxal (MGO) largely accumulates in the circulation due to diabetes. We addressed whether macrophages exposed to MGO exhibited damaging effect on the coronary artery and whether urocortin2 (UCN2) serve as protecting factors against such diabetes-associated complication. Type 2 diabetes was induced by high-fat diet and a single low-dose streptozotocin in mice. Small extracellular vesicles (sEV) derived from MGO-treated macrophages (MGO-sEV) were used to produce diabetes-like CMD. UCN2 was examined for a protective role against CMD. The involvement of arginase1 and IL-33 was tested by pharmacological inhibitor and IL-33-/- mice. MGO-sEV was capable of causing coronary artery endothelial dysfunction similar to that by diabetes. Immunocytochemistry studies of diabetic coronary arteries supported the transfer of arginase1 from macrophages to endothelial cells. Mechanism studies revealed arginase1 contributed to the impaired endothelium-dependent relaxation of coronary arteries in diabetic and MGO-sEV-treated mice. UCN2 significantly improved coronary artery endothelial function, and prevented MGO elevation in diabetic mice or enrichment of arginase1 in MGO-sEV. Diabetes caused a reduction of IL-33, which was also reversed by UCN2. IL-33-/- mice showed impaired endothelium-dependent relaxation of coronary arteries, which can be mitigated by arginase1 inhibition but can't be improved by UCN2 anymore, indicating the importance of restoring IL-33 for the protection against diabetic CMD by UCN2. Our data suggest that MGO-sEV induces CMD via shuttling arginase1 to coronary arteries. UCN2 is able to protect against diabetic CMD via modulating MGO-altered macrophage sEV cargoes.


Subject(s)
Diabetes Complications , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Urocortins , Animals , Humans , Mice , Diabetes Mellitus, Experimental/drug therapy , Endothelial Cells , Interleukin-33 , Macrophages , Magnesium Oxide/pharmacology , Urocortins/genetics
20.
ACS Biomater Sci Eng ; 10(1): 537-549, 2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38065085

ABSTRACT

Commercially available guided bone regeneration (GBR) membranes often exhibit limited mechanical properties or bioactivity, leading to poor performance in repairing bone defects. To surmount this limitation, we developed a Janus structural composite membrane (Mg-MgO/PCL) reinforced by dual Mg (Mg sheets and MgO NPs) by using a combined processing technique involving casting and electrospinning. Results showed that the addition of Mg sheets and MgO NPs enhanced the mechanical properties of the composite membrane for osteogenic space maintenance, specifically tensile strength (from 10.2 ± 1.2 to 50.3 ± 4.5 MPa) and compression force (from 0 to 0.94 ± 0.09 N mm-1), through Mg sheet reinforcement and improved crystallization. The dense cast side of the Janus structure membrane displayed better fibroblast barrier capacity than a single fiber structure; meanwhile, the PCL matrix protected the Mg sheet from severe corrosion due to predeformation. The porous microfibers side supported preosteoblast cell adhesion, enhanced osteogenesis, and angiogenesis in vitro, through the biomimetic extracellular matrix and sustainable Mg2+ release. Furthermore, the Mg-MgO/PCL membrane incorporating 2 wt % MgO NPs exhibited remarkable antimicrobial properties, inducing over 88.75% apoptosis in Staphylococcus aureus. An in vivo experiment using the rat skull defect model (Φ = 5 mm) confirmed that the Mg-MgO/PCL membrane significantly improved new bone formation postsurgery. Collectively, our investigation provides valuable insights into the design of multifunctional membranes for clinical oral GBR application.


Subject(s)
Magnesium Oxide , Polyesters , Rats , Animals , Magnesium Oxide/pharmacology , Polyesters/pharmacology , Polyesters/chemistry , Bone Regeneration , Osteogenesis , Cell Adhesion
SELECTION OF CITATIONS
SEARCH DETAIL